A Precisely Positioned MED12 Activation Helix Stimulates CDK8 Kinase Activity

Total Page:16

File Type:pdf, Size:1020Kb

A Precisely Positioned MED12 Activation Helix Stimulates CDK8 Kinase Activity A precisely positioned MED12 activation helix stimulates CDK8 kinase activity Felix Klatta, Alexander Leitnerb, Iana V. Kima, Hung Ho-Xuanc, Elisabeth V. Schneiderd,e, Franziska Langhammera, Robin Weinmanna, Melanie R. Müllera, Robert Huberd,f,g,1, Gunter Meisterc, and Claus-D. Kuhna,1 aGene Regulation by Non-Coding RNA, Elite Network of Bavaria and University of Bayreuth, 95447 Bayreuth, Germany; bDepartment of Biology, Institute of Molecular Systems Biology, Eidgenössiche Technische Hochschule Zürich, 8093 Zürich, Switzerland; cBiochemistry Center Regensburg, Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany; dMax Planck Institute of Biochemistry, 82152 Martinsried, Germany; eProteros Biostructures GmbH, 82152 Martinsried, Germany; fCenter of Medical Biotechnology, University of Duisburg-Essen, 45117 Essen, Germany; and gDepartment of Chemistry, Technical University of Munich, 85747 Garching, Germany Contributed by Robert Huber, December 21, 2019 (sent for review October 10, 2019; reviewed by Patrick Cramer and Matthias Geyer) The Mediator kinase module regulates eukaryotic transcription by with altered subunit composition accompanied by a potentially phosphorylating transcription-related targets and by modulating altered activity and target specificity (14). CDK8 and its paralog the association of Mediator and RNA polymerase II. The activity of its CDK19 were found to phosphorylate a wide variety of substrates catalytic core, cyclin-dependent kinase 8 (CDK8), is controlled by involved in transcription, DNA repair, and metabolic processes Cyclin C and regulatory subunit MED12, with its deregulation (11). This wealth of substrates likely lies at the heart of the context- contributing to numerous malignancies. Here, we combine in vitro dependent role of the Mediator kinase module in metazoan tran- biochemistry, cross-linking coupled to mass spectrometry, and scription. Moreover, the central role of CDK8 in transcription in vivo studies to describe the binding location of the N-terminal regulation explains why it is a potent oncogene that is involved in segment of MED12 on the CDK8/Cyclin C complex and to gain the progression of malignant cancers, like colorectal cancer and mechanistic insights into the activation of CDK8 by MED12. Our data acute myeloid leukemia (15–17). Due to its oncogenic potential, demonstrate that the N-terminal portion of MED12 wraps around the effective and selective inhibition of CDK8 is of great interest to CDK8, whereby it positions an “activation helix” close to the T-loop drug development and therefore spurred significant efforts to de- of CDK8 for its activation. Intriguingly, mutations in the activation velop CDK8-specific inhibitors. Despite the availability of structural helix that are frequently found in cancers do not diminish the affinity information on CDK8 bound to Cyclin C (18), the development of BIOCHEMISTRY of MED12 for CDK8, yet likely alter the exact positioning of the CDK8-specific inhibitors is hampered by the to date unclear activation helix. Furthermore, we find the transcriptome-wide mechanism of CDK8 activation. In contrast to the common acti- gene-expression changes in human cells that result from a mutation vation mechanism of cyclin-dependent kinases, CDK8 does not in the MED12 activation helix to correlate with deregulated genes in require phosphorylation of its T-loop for full activity. Instead, breast and colon cancer. Finally, functional assays in the presence of CDK8 T-loop phosphorylation seems to be functionally replaced by kinase inhibitors reveal that binding of MED12 remodels the active MED12 binding to CDK8, which results in its activation (5, 19). site of CDK8 and thereby precludes the inhibition of ternary CDK8 complexes by type II kinase inhibitors. Taken together, our results Significance not only allow us to propose a revised model of how CDK8 activity is regulated by MED12, but also offer a path forward in developing small molecules that target CDK8 in its MED12-bound form. CDK8, the catalytic core of the Mediator kinase module, is activated by MED12. We describe the architecture of the ternary complex of CDK8 | MED12 | Mediator kinase module | Mediator | kinase inhibitors CDK8, Cyclin C, and MED12, and we decipher the mechanism by which MED12 activates CDK8. We find the N-terminal segment of MED12towraparoundtheCDK8moleculetoplacean“activation ediator is essential for all RNA polymerase II (Pol II)- helix” in proximity of its T-loop. Exact placement of the helix, not Mmediated transcription in eukaryotes because it allows for MED12 binding alone, is crucial for CDK8 activation. Moreover, the communication between enhancer-bound transcription factors – MED12 binding precludes binding of type II kinase inhibitors to the and Pol II (1 4). In humans Mediator comprises 30 protein subunits active site of CDK8. As many cancer patients carry mutations in the that are subdivided into four modules: The head, the middle, the tail, MED12 activation helix, our study invigorates the development of and the kinase module. Whereas the first three modules form a CDK8-specific compounds as well as offers functional insights into complex that is often referred to as the Mediator complex, the kinase the Mediator kinase module. module only reversibly associates with the three-module Mediator – complex (5 7). Structural information on the isolated Mediator Author contributions: F.K. and C.-D.K. designed research; F.K., A.L., I.V.K., H.H.-X., E.V.S., complex and on the head and the middle module of Mediator, F.L., R.W., and M.R.M. performed research; G.M. and C.-D.K. supervised the study; F.K., collectively termed core Mediator, bound to Pol II provided first A.L., I.V.K., E.V.S., and R.H. analyzed data; and F.K., R.H., G.M., and C.-D.K. wrote insights into how Mediator is able to activate Pol II (8, 9). In contrast the paper. to the Mediator complex, the structure of the kinase module is un- Reviewers: P.C., Max Planck Institute for Biophysical Chemistry; and M.G., University known and its role during the transcription cycle is unclear. Despite of Bonn. the fact that, in yeast, the three-module Mediator complex is unable The authors declare no competing interest. to simultaneously bind Pol II and the kinase module (10), deletion or Published under the PNAS license. inhibitor of CDK8 results only in minor gene-expression changes (11, Data deposition: The mass spectrometry data were deposited to the ProteomeXchange 12). However, using human proteins in a mostly recombinant in vitro Consortium via the PRIDE partner repository (identifier PXD015394). The structure of Compound A in complex with CDK8 (1–403)/Cyclin C were deposited to the Protein Data system, the kinase module was found to repress transcription by Bank (PDB ID code 6T41). All sequencing data been deposited in the Gene Expression inhibiting Mediator coactivator function (13). Omnibus (GEO) database, https://www.ncbi.nlm.nih.gov/geo (accession no. GSE135458). The Mediator kinase module consists of four subunits: Cyclin- 1To whom correspondence may be addressed. Email: [email protected] or claus. dependent kinase 8 (CDK8), its corresponding cyclin, Cyclin C, and [email protected]. two large regulatory subunits, MED12 and MED13. Moreover, in This article contains supporting information online at https://www.pnas.org/lookup/suppl/ metazoans all kinase module subunits, except for Cyclin C, possess doi:10.1073/pnas.1917635117/-/DCSupplemental. paralogues that can be utilized for the assembly of kinase modules www.pnas.org/cgi/doi/10.1073/pnas.1917635117 PNAS Latest Articles | 1of12 Downloaded by guest on September 27, 2021 The current model for CDK8 activation proposes that the N-terminal necessary for CDK8 activation. In addition, we find that disease- segmentofMED12bindstoasurfacegrooveonCyclinC,andthereby causing MED12 mutations likely lead to mis-positioning of the enhances CDK8 activity (19). However, how MED12 binding to activation helix, and thereby abrogate CDK8 activity. In further this distant surface groove could be coupled to repositioning of support, we show that the transcriptome-wide gene expression the CDK8 T-loop remains unclear and the current model is changes in HCT116 cells that carry a MED12 E33Q mutation therefore controversial. phenocopy multiple cancer-related transcriptome alterations. Fi- To investigate how MED12 activates CDK8, we established a nally, we demonstrate that, in contrast to type I inhibitors, type II recombinant expression and purification scheme that resulted in kinase inhibitors lose much of their inhibitory potential when used monodisperse, highly pure ternary CDK8/Cyclin C/MED12 against ternary MED12-bound CDK8/Cyclin C in vitro. Taken complexes. Using these complexes, we carried out chemical cross- together, our results establish that an activation helix in MED12 linking coupled to mass spectrometry. Unexpectedly, the resulting functionally replaces T-loop phosphorylation of CDK8. Further- lysine- and aspartate/glutamate-mediated cross-links suggest a more, MED12 binding likely remodels the CDK8 active site, thus substantially revised mechanism for how the N-terminal portion of preventing type II kinase inhibitors from efficiently targeting MED12 binds and activates CDK8: MED12 makes extensive CDK8 bound by MED12 in vivo. contacts to CDK8 with no crucial contacts to Cyclin C. In- triguingly, MED12 thereby places a critical “activation helix” in Results direct vicinity of the T-loop of CDK8, leading to its conformational MED12 Binding Stabilizes
Recommended publications
  • Methylation Status of Vitamin D Receptor Gene Promoter in Adrenocortical Carcinoma
    UNIVERSITÀ DEGLI STUDI DI PADOVA DEPARTMENT OF CARDIAC, THORACIC AND VASCULAR SCIENCES Ph.D Course Medical Clinical and Experimental Sciences Curriculum Clinical Methodology, Endocrinological, Diabetological and Nephrological Sciences XXIX° SERIES METHYLATION STATUS OF VITAMIN D RECEPTOR GENE PROMOTER IN ADRENOCORTICAL CARCINOMA Coordinator: Ch.mo Prof. Annalisa Angelini Supervisor: Ch.mo Prof. Francesco Fallo Ph.D Student: Andrea Rebellato TABLE OF CONTENTS SUMMARY 3 INTRODUCTION 4 PART 1: ADRENOCORTICAL CARCINOMA 4 1.1 EPIDEMIOLOGY 4 1.2 GENETIC PREDISPOSITION 4 1.3 CLINICAL PRESENTATION 6 1.4 DIAGNOSTIC WORK-UP 7 1.4.1 Biochemistry 7 1.4.2 Imaging 9 1.5 STAGING 10 1.6 PATHOLOGY 11 1.7 MOLECULAR PATHOLOGY 14 1.7.1 DNA content 15 1.7.2 Chromosomal aberrations 15 1.7.3 Differential gene expression 16 1.7.4 DNA methylation 17 1.7.5 microRNAs 18 1.7.6 Gene mutations 19 1.8 PATHOPHYSIOLOGY OF MOLECULAR SIGNALLING 21 PATHWAYS 1.8.1 IGF-mTOR pathway 21 1.8.2 WNTsignalling pathway 22 1.8.3 Vascular endothelial growth factor 23 1.9 THERAPY 24 1.9.1 Surgery 24 1.9.2 Adjuvant Therapy 27 1.9.2.1 Mitotane 27 1.9.2.2 Cytotoxic chemotherapy 30 1.9.2.3 Targeted therapy 31 1.9.2.4 Therapy for hormone excess 31 1.9.2.5 Radiation therapy 32 1.9.2.6 Other local therapies 32 1.10 PROGNOSTIC FACTORS AND PREDICTIVE MARKERS 32 PART 2: VITAMIN D 35 2.1 VITAMIN D AND ITS BIOACTIVATION 35 2.2 THE VITAMIN D RECEPTOR 37 2.3 GENOMIC MECHANISM OF 1,25(OH)2D3-VDR COMPLEX 38 2.4 CLASSICAL ROLES OF VITAMIN D 40 2.4.1 Intestine 40 2.4.2 Kidney 41 2.4.3 Bone 41 2.5 PLEIOTROPIC
    [Show full text]
  • Evidence for Differential Alternative Splicing in Blood of Young Boys With
    Stamova et al. Molecular Autism 2013, 4:30 http://www.molecularautism.com/content/4/1/30 RESEARCH Open Access Evidence for differential alternative splicing in blood of young boys with autism spectrum disorders Boryana S Stamova1,2,5*, Yingfang Tian1,2,4, Christine W Nordahl1,3, Mark D Shen1,3, Sally Rogers1,3, David G Amaral1,3 and Frank R Sharp1,2 Abstract Background: Since RNA expression differences have been reported in autism spectrum disorder (ASD) for blood and brain, and differential alternative splicing (DAS) has been reported in ASD brains, we determined if there was DAS in blood mRNA of ASD subjects compared to typically developing (TD) controls, as well as in ASD subgroups related to cerebral volume. Methods: RNA from blood was processed on whole genome exon arrays for 2-4–year-old ASD and TD boys. An ANCOVA with age and batch as covariates was used to predict DAS for ALL ASD (n=30), ASD with normal total cerebral volumes (NTCV), and ASD with large total cerebral volumes (LTCV) compared to TD controls (n=20). Results: A total of 53 genes were predicted to have DAS for ALL ASD versus TD, 169 genes for ASD_NTCV versus TD, 1 gene for ASD_LTCV versus TD, and 27 genes for ASD_LTCV versus ASD_NTCV. These differences were significant at P <0.05 after false discovery rate corrections for multiple comparisons (FDR <5% false positives). A number of the genes predicted to have DAS in ASD are known to regulate DAS (SFPQ, SRPK1, SRSF11, SRSF2IP, FUS, LSM14A). In addition, a number of genes with predicted DAS are involved in pathways implicated in previous ASD studies, such as ROS monocyte/macrophage, Natural Killer Cell, mTOR, and NGF signaling.
    [Show full text]
  • The Role of Gli3 in Inflammation
    University of New Hampshire University of New Hampshire Scholars' Repository Doctoral Dissertations Student Scholarship Winter 2020 THE ROLE OF GLI3 IN INFLAMMATION Stephan Josef Matissek University of New Hampshire, Durham Follow this and additional works at: https://scholars.unh.edu/dissertation Recommended Citation Matissek, Stephan Josef, "THE ROLE OF GLI3 IN INFLAMMATION" (2020). Doctoral Dissertations. 2552. https://scholars.unh.edu/dissertation/2552 This Dissertation is brought to you for free and open access by the Student Scholarship at University of New Hampshire Scholars' Repository. It has been accepted for inclusion in Doctoral Dissertations by an authorized administrator of University of New Hampshire Scholars' Repository. For more information, please contact [email protected]. THE ROLE OF GLI3 IN INFLAMMATION BY STEPHAN JOSEF MATISSEK B.S. in Pharmaceutical Biotechnology, Biberach University of Applied Sciences, Germany, 2014 DISSERTATION Submitted to the University of New Hampshire in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy In Biochemistry December 2020 This dissertation was examined and approved in partial fulfillment of the requirement for the degree of Doctor of Philosophy in Biochemistry by: Dissertation Director, Sherine F. Elsawa, Associate Professor Linda S. Yasui, Associate Professor, Northern Illinois University Paul Tsang, Professor Xuanmao Chen, Assistant Professor Don Wojchowski, Professor On October 14th, 2020 ii ACKNOWLEDGEMENTS First, I want to express my absolute gratitude to my advisor Dr. Sherine Elsawa. Without her help, incredible scientific knowledge and amazing guidance I would not have been able to achieve what I did. It was her encouragement and believe in me that made me overcome any scientific struggles and strengthened my self-esteem as a human being and as a scientist.
    [Show full text]
  • COFACTORS of the P65- MEDIATOR COMPLEX
    COFACTORS OF THE April 5 p65- MEDIATOR 2011 COMPLEX Honors Thesis Department of Chemistry and Biochemistry NICHOLAS University of Colorado at Boulder VICTOR Faculty Advisor: Dylan Taatjes, PhD PARSONNET Committee Members: Rob Knight, PhD; Robert Poyton, PhD Table of Contents Abstract ......................................................................................................................................................... 3 Introduction .................................................................................................................................................. 4 The Mediator Complex ............................................................................................................................. 6 The NF-κB Transcription Factor ................................................................................................................ 8 Hypothesis............................................................................................................................................... 10 Results ......................................................................................................................................................... 11 Discussion.................................................................................................................................................... 15 p65-only factors ...................................................................................................................................... 16 p65-enriched factors ..............................................................................................................................
    [Show full text]
  • MED12 Related Disorders FTNW
    MED12 related disorders rarechromo.org What are the MED12 related disorders? MED12 related disorders are a group of disorders that primarily affect boys. Most boys with MED12 related disorders have intellectual disability/ developmental delay, behavioural problems and low muscle tone. MED12 related disorders occur when the MED12 gene has lost its normal function. Genes are instructions which have important roles in our growth and development. They are made of DNA and are incorporated into organised structures called chromosomes. Chromosomes therefore contain our genetic information. Chromosomes are located in our cells, the building blocks of our bodies. The MED12 gene is located on the X chromosome. The X chromosome is one of the sex chromosomes that determine a person’s gender. Men have one X chromosome and one Y chromosome, while women have two X chromosomes. Because the MED12 gene is located on the X chromosome, men have only one copy of the gene, while women have two copies. A change in the MED12 gene can cause symptoms in men. Women with a change in the MED12 gene usually have no symptoms. They carry a second copy of the gene that does function normally. Women may show mild features such as learning difficulties. The MED12 related disorders are FG syndrome , Lujan syndrome and the X- linked recessive form of Ohdo syndrome . Although these syndromes differ, they also have the overlapping features listed on page 2. The way that these conditions are inherited is called ‘X-linked’ or ‘X-linked’ recessive. In 2007 it was discovered that changes in the MED12 gene were responsible for the symptoms and features in several boys with FG syndrome.
    [Show full text]
  • Regulatory Functions of the Mediator Kinases CDK8 and CDK19 Charli B
    TRANSCRIPTION 2019, VOL. 10, NO. 2, 76–90 https://doi.org/10.1080/21541264.2018.1556915 REVIEW Regulatory functions of the Mediator kinases CDK8 and CDK19 Charli B. Fant and Dylan J. Taatjes Department of Biochemistry, University of Colorado, Boulder, CO, USA ABSTRACT ARTICLE HISTORY The Mediator-associated kinases CDK8 and CDK19 function in the context of three additional Received 19 September 2018 proteins: CCNC and MED12, which activate CDK8/CDK19 kinase function, and MED13, which Revised 13 November 2018 enables their association with the Mediator complex. The Mediator kinases affect RNA polymerase Accepted 20 November 2018 II (pol II) transcription indirectly, through phosphorylation of transcription factors and by control- KEYWORDS ling Mediator structure and function. In this review, we discuss cellular roles of the Mediator Mediator kinase; enhancer; kinases and mechanisms that enable their biological functions. We focus on sequence-specific, transcription; RNA DNA-binding transcription factors and other Mediator kinase substrates, and how CDK8 or CDK19 polymerase II; chromatin may enable metabolic and transcriptional reprogramming through enhancers and chromatin looping. We also summarize Mediator kinase inhibitors and their therapeutic potential. Throughout, we note conserved and divergent functions between yeast and mammalian CDK8, and highlight many aspects of kinase module function that remain enigmatic, ranging from potential roles in pol II promoter-proximal pausing to liquid-liquid phase separation. Introduction and MED13L associate in a mutually exclusive fash- ion with MED12 and MED13 [12], and their poten- The CDK8 kinase exists in a 600 kDa complex tial functional distinctions remain unclear. known as the CDK8 module, which consists of four CDK8 is considered both an oncogene [13–15] proteins (CDK8, CCNC, MED12, MED13).
    [Show full text]
  • MED12 Mutations Link Intellectual Disability Syndromes with Dysregulated GLI3-Dependent Sonic Hedgehog Signaling
    MED12 mutations link intellectual disability syndromes with dysregulated GLI3-dependent Sonic Hedgehog signaling Haiying Zhoua, Jason M. Spaethb, Nam Hee Kimb, Xuan Xub, Michael J. Friezc, Charles E. Schwartzc, and Thomas G. Boyerb,1 aDepartment of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, CA 94270; bDepartment of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; and cGreenwood Genetic Center, Greenwood, SC 29646 Edited by Robert G. Roeder, The Rockefeller University, New York, NY, and approved September 13, 2012 (received for review December 20, 2011) Recurrent missense mutations in the RNA polymerase II Mediator divergent “tail” and “kinase” modules through which most activa- subunit MED12 are associated with X-linked intellectual disability tors and repressors target Mediator. The kinase module, compris- (XLID) and multiple congenital anomalies, including craniofacial, ing MED12, MED13, CDK8, and cyclin C, has been ascribed both musculoskeletal, and behavioral defects in humans with FG (or activating and repressing functions and exists in variable association Opitz-Kaveggia) and Lujan syndromes. However, the molecular with Mediator, implying that its regulatory role is restricted to a mechanism(s) underlying these phenotypes is poorly understood. subset of RNA polymerase II transcribed genes. Here we report that MED12 mutations R961W and N1007S causing Within the Mediator kinase module, MED12 is a critical trans- FG and Lujan syndromes, respectively, disrupt a Mediator-imposed ducer of regulatory information conveyed by signal-activated transcription factors linked to diverse developmental pathways, constraint on GLI3-dependent Sonic Hedgehog (SHH) signaling. We including the EGF, Notch, Wnt, and Hedgehog pathways (10–12).
    [Show full text]
  • MED12 Gene Mediator Complex Subunit 12
    MED12 gene mediator complex subunit 12 Normal Function The MED12 gene provides instructions for making a protein called mediator complex subunit 12. As its name suggests, this protein forms one part (subunit) of the mediator complex, which is a group of about 25 proteins that work together to regulate gene activity. The mediator complex physically links transcription factors, which are proteins that influence whether genes are turned on or off, with an enzyme called RNA polymerase II. Once transcription factors are attached, this enzyme initiates gene transcription, the process by which information stored in a gene's DNA is used to build proteins. Researchers believe that the MED12 protein is involved in many aspects of early development, including the development of nerve cells (neurons) in the brain. The MED12 protein is part of several chemical signaling pathways within cells. These pathways help direct a broad range of cellular activities, such as cell growth, cell movement (migration), and the process by which cells mature to carry out specific functions (differentiation). Health Conditions Related to Genetic Changes FG syndrome At least two mutations in the MED12 gene have been found to cause FG syndrome, which is characterized by intellectual disability, behavioral problems, and physical abnormalities including weak muscle tone (hypotonia) and obstruction of the anal opening (imperforate anus). The mutations that cause FG syndrome each change a single protein building block ( amino acid) in the MED12 protein. One mutation replaces the amino acid arginine with the amino acid tryptophan at protein position 961 (written as Arg961Trp or R961W). The other replaces the amino acid glycine with the amino acid glutamic acid at protein position 958 (written as Gly958Glu or G958E).
    [Show full text]
  • The Role of Med12 in Tumorigenesis
    Helsinki Graduate Program in Biotechnology and Molecular Biology (GPBM)/ Integrative Life Sciences (ILS) Doctoral Program THE ROLE OF MED12 IN TUMORIGENESIS Kati Kämpjärvi Department of Medical and Clinical Genetics, Medicum & Genome-Scale Biology Research Program, Research Programs Unit Faculty of Medicine University of Helsinki Helsinki, Finland Academic dissertation To be publicly discussed, with the permission of the Faculty of Medicine, University of Helsinki, in Haartman Institute, Lecture Hall 1, Haartmaninkatu 3, Helsinki, on the 18th of November 2016, at 12 noon. Helsinki 2016 Supervised by Docent Pia Vahteristo, Ph.D. Department of Medical and Clinical Genetics, Medicum Genome-Scale Biology Research Program, Research Programs Unit Faculty of Medicine University of Helsinki Helsinki, Finland Academy Professor Lauri A. Aaltonen, M.D., Ph.D. Department of Medical and Clinical Genetics, Medicum Genome-Scale Biology Research Program, Research Programs Unit Faculty of Medicine University of Helsinki Helsinki, Finland Reviewed by Docent Miina Ollikainen, Ph.D. Institute for Molecular Medicine Finland, FIMM Department of Public Health, Clinicum Faculty of Medicine University of Helsinki Helsinki, Finland Docent Katri Pylkäs, Ph.D. Laboratory of Cancer Genetics and Tumor Biology Biocenter Oulu Faculty of Medicine University of Oulu Oulu, Finland Official opponent Professor Anne Kallioniemi, M.D., Ph.D. BioMediTech University of Tampere Tampere, Finland ISBN 978-951-51-2643-6 (paperback) ISBN 978-951-51-2644-3 (PDF) htpp://ethesis.helsinki.fi
    [Show full text]
  • The MED12-MED13 Module of Mediator Regulates the Timing of Embryo Patterning in Arabidopsis C
    RESEARCH ARTICLE 113 Development 137, 113-122 (2010) doi:10.1242/dev.043174 The MED12-MED13 module of Mediator regulates the timing of embryo patterning in Arabidopsis C. Stewart Gillmor1, Mee Yeon Park1, Michael R. Smith1, Robert Pepitone2, Randall A. Kerstetter2 and R. Scott Poethig1,* SUMMARY The Arabidopsis embryo becomes patterned into central and peripheral domains during the first few days after fertilization. A screen for mutants that affect this process identified two genes, GRAND CENTRAL (GCT)andCENTER CITY (CCT). Mutations in GCT and CCT delay the specification of central and peripheral identity and the globular-to-heart transition, but have little or no effect on the initial growth rate of the embryo. Mutant embryos eventually recover and undergo relatively normal patterning, albeit at an inappropriate size. GCT and CCT were identified as the Arabidopsis orthologs of MED13 and MED12 – evolutionarily conserved proteins that act in association with the Mediator complex to negatively regulate transcription. The predicted function of these proteins combined with the effect of gct and cct on embryo development suggests that MED13 and MED12 regulate pattern formation during Arabidopsis embryogenesis by transiently repressing a transcriptional program that interferes with this process. Their mutant phenotype reveals the existence of a previously unknown temporal regulatory mechanism in plant embryogenesis. KEY WORDS: Arabidopsis, Embryo, Heterochrony, MEDIATOR, MED12, MED13, Polarity, KANADI INTRODUCTION (McConnell and Barton, 1998).
    [Show full text]
  • Identification of MED12 and RBM10 As Novel Thyroid Cancer Genes Associated with Tumor Virulence
    Published OnlineFirst June 20, 2017; DOI: 10.1158/1078-0432.CCR-17-1183 Biology of Human Tumors Clinical Cancer Research Genomic Alterations in Fatal Forms of Non-Anaplastic Thyroid Cancer: Identification of MED12 and RBM10 as Novel Thyroid Cancer Genes Associated with Tumor Virulence Tihana Ibrahimpasic1,2, Bin Xu3,Inigo~ Landa1, Snjezana Dogan3, Sumit Middha3, Venkatraman Seshan4, Shyam Deraje1, Diane L. Carlson5, Jocelyn Migliacci2, Jeffrey A. Knauf1, Brian Untch1, Michael F. Berger1,3, Luc Morris1,2, R. Michael Tuttle6, Timothy Chan1, James A. Fagin1,6, Ronald Ghossein3, and Ian Ganly1,2 Abstract Purpose: Patients with anaplastic thyroid cancer (ATC) have a were identified. There was a high frequency of mutations in very high death rate. In contrast, deaths from non-anaplastic MED12 and these were mutually exclusive to TERT promoter thyroid (NAT) cancer are much less common. The genetic altera- mutations and also to BRAF and RAS mutations. In addition, a tions in fatal NAT cancers have not been reported. high frequency of mutations in RBM10 was identified and these Experimental Design: We performed next-generation sequenc- co-occurred with RAS mutations and PIK3CA mutations. Com- ing of 410 cancer genes from 57 fatal NAT primary cancers. Results pared with the PTCs in TCGA, there were higher frequencies of were compared with The Cancer Genome Atlas study (TCGA mutations in TP53, POLE, PI3K/AKT/mTOR pathway effectors, study) of papillary thyroid cancers (PTCs) and to the genomic SWI/SNF subunits, and histone methyltransferases. changes reported in ATC. Conclusions: These data support a model, whereby fatal NAT Results: There was a very high prevalence of TERT promoter cancers arise from well-differentiated tumors through the accu- mutations, comparable with that of ATC, and these co-occurred mulation of key additional genetic abnormalities.
    [Show full text]
  • Mutations in MED12 Cause X-Linked Ohdo Syndrome
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector REPORT Mutations in MED12 Cause X-Linked Ohdo Syndrome Anneke T. Vulto-van Silfhout,1,2 Bert B.A. de Vries,1,2 Bregje W.M. van Bon,1,2 Alexander Hoischen,1,2,3 Martina Ruiterkamp-Versteeg,1 Christian Gilissen,1,2,3 Fangjian Gao,4 Marloes van Zwam,5 Cornelis L. Harteveld,6 Anthonie J. van Essen,7 Ben C.J. Hamel,1 Tjitske Kleefstra,1,2 Miche`l A.A.P. Willemsen,8,9 Helger G. Yntema,1 Hans van Bokhoven,1,9,10 Han G. Brunner,1,2,3,* Thomas G. Boyer,4 and Arjan P.M. de Brouwer1,9,10 Ohdo syndrome comprises a heterogeneous group of disorders characterized by intellectual disability (ID) and typical facial features, including blepharophimosis. Clinically, these blepharophimosis-ID syndromes have been classified in five distinct subgroups, including the Maat-Kievit-Brunner (MKB) type, which, in contrast to the others, is characterized by X-linked inheritance and facial coarsening at older age. We performed exome sequencing in two families, each with two affected males with Ohdo syndrome MKB type. In the two families, MED12 missense mutations (c.3443G>A [p.Arg1148His] or c.3493T>C [p.Ser1165Pro]) segregating with the phenotype were identified. Upon subsequent analysis of an additional cohort of nine simplex male individuals with Ohdo syndrome, one additional de novo missense change (c.5185C>A [p.His1729Asn]) in MED12 was detected. The occurrence of three different hemizygous missense mutations in three unrelated families affected by Ohdo syndrome MKB type shows that mutations in MED12 are the underlying cause of this X-linked form of Ohdo syndrome.
    [Show full text]